1
|
Portillo AL, Rojas EA, Mehboob M, Moinuddin A, Balint E, Feng E, Silvestri C, Vahedi F, Ritchie TM, Mansour AJ, Bramson JL, Ashkar AA. CD56 does not contribute to the antitumor, tissue homing, and glycolytic capacity of human NK cells. J Leukoc Biol 2025; 117:qiae227. [PMID: 39449625 DOI: 10.1093/jleuko/qiae227] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Accepted: 10/23/2024] [Indexed: 10/26/2024] Open
Abstract
Natural killer (NK) cells are critical innate immune cells involved in the clearance of virally infected and malignant cells. Human NK cells are distinguished by their surface expression of CD56 and a lack of CD3. While CD56 expression and cell surface density has long been used as the prototypic marker to characterize primary human NK cell functional subsets, the exact functional role of CD56 in primary human NK cells is still not fully understood. Here, we eliminated the expression of CD56 in human ex vivo expanded NK cells (CD56bright) using CRISPR/Cas9 in order to assess the function of CD56 in this highly activated and cytotoxic NK cell population. We show that the expression of CD56 has no effect on NK cell proliferative capacity or expression of various activation and inhibitory markers. Further, CD56 does not contribute to NK cell-mediated cytotoxicity, inflammatory cytokine production, or the ability of NK cells to control tumor engraftment in vivo. We also found that while deletion of CD56 did not impact NK cell glycolytic metabolism, it did increase NK cell reliance on oxidative phosphorylation. Last, CD56 does not alter expanded NK cell in vivo tissue trafficking. Our results indicate that while CD56 expression could be used to indicate a hyperfunctional state of NK cells, it does not directly influence the antitumor functions of expanded NK cells.
Collapse
Affiliation(s)
- Ana L Portillo
- Department of Medicine, McMaster University, McMaster Children's Hospital, 1200 Main Street West, Hamilton, ON L8N 3Z5, Canada
- McMaster Immunology Research Centre, McMaster University, MDCL 4010, 1280 Main Street West, Hamilton, ON L8S 4K1, Canada
- Centre for Discovery in Cancer Research, McMaster University, MDCL 5106, 1280 Main Street West, Hamilton, ON L8S 4M1, Canada
| | - Eduardo A Rojas
- Department of Medicine, McMaster University, McMaster Children's Hospital, 1200 Main Street West, Hamilton, ON L8N 3Z5, Canada
- McMaster Immunology Research Centre, McMaster University, MDCL 4010, 1280 Main Street West, Hamilton, ON L8S 4K1, Canada
| | - Misaal Mehboob
- Department of Medicine, McMaster University, McMaster Children's Hospital, 1200 Main Street West, Hamilton, ON L8N 3Z5, Canada
- McMaster Immunology Research Centre, McMaster University, MDCL 4010, 1280 Main Street West, Hamilton, ON L8S 4K1, Canada
- Centre for Discovery in Cancer Research, McMaster University, MDCL 5106, 1280 Main Street West, Hamilton, ON L8S 4M1, Canada
| | - Adnan Moinuddin
- Department of Medicine, McMaster University, McMaster Children's Hospital, 1200 Main Street West, Hamilton, ON L8N 3Z5, Canada
- McMaster Immunology Research Centre, McMaster University, MDCL 4010, 1280 Main Street West, Hamilton, ON L8S 4K1, Canada
- Centre for Discovery in Cancer Research, McMaster University, MDCL 5106, 1280 Main Street West, Hamilton, ON L8S 4M1, Canada
| | - Elizabeth Balint
- Department of Medicine, McMaster University, McMaster Children's Hospital, 1200 Main Street West, Hamilton, ON L8N 3Z5, Canada
- McMaster Immunology Research Centre, McMaster University, MDCL 4010, 1280 Main Street West, Hamilton, ON L8S 4K1, Canada
- Centre for Discovery in Cancer Research, McMaster University, MDCL 5106, 1280 Main Street West, Hamilton, ON L8S 4M1, Canada
| | - Emily Feng
- Department of Medicine, McMaster University, McMaster Children's Hospital, 1200 Main Street West, Hamilton, ON L8N 3Z5, Canada
- McMaster Immunology Research Centre, McMaster University, MDCL 4010, 1280 Main Street West, Hamilton, ON L8S 4K1, Canada
- Centre for Discovery in Cancer Research, McMaster University, MDCL 5106, 1280 Main Street West, Hamilton, ON L8S 4M1, Canada
| | - Christopher Silvestri
- Department of Medicine, McMaster University, McMaster Children's Hospital, 1200 Main Street West, Hamilton, ON L8N 3Z5, Canada
- McMaster Immunology Research Centre, McMaster University, MDCL 4010, 1280 Main Street West, Hamilton, ON L8S 4K1, Canada
- Centre for Discovery in Cancer Research, McMaster University, MDCL 5106, 1280 Main Street West, Hamilton, ON L8S 4M1, Canada
| | - Fatemeh Vahedi
- Department of Medicine, McMaster University, McMaster Children's Hospital, 1200 Main Street West, Hamilton, ON L8N 3Z5, Canada
- McMaster Immunology Research Centre, McMaster University, MDCL 4010, 1280 Main Street West, Hamilton, ON L8S 4K1, Canada
- Centre for Discovery in Cancer Research, McMaster University, MDCL 5106, 1280 Main Street West, Hamilton, ON L8S 4M1, Canada
| | - Tyrah M Ritchie
- Department of Medicine, McMaster University, McMaster Children's Hospital, 1200 Main Street West, Hamilton, ON L8N 3Z5, Canada
- McMaster Immunology Research Centre, McMaster University, MDCL 4010, 1280 Main Street West, Hamilton, ON L8S 4K1, Canada
| | - Alexa J Mansour
- Department of Medicine, McMaster University, McMaster Children's Hospital, 1200 Main Street West, Hamilton, ON L8N 3Z5, Canada
- McMaster Immunology Research Centre, McMaster University, MDCL 4010, 1280 Main Street West, Hamilton, ON L8S 4K1, Canada
- Centre for Discovery in Cancer Research, McMaster University, MDCL 5106, 1280 Main Street West, Hamilton, ON L8S 4M1, Canada
| | - Jonathan L Bramson
- Department of Medicine, McMaster University, McMaster Children's Hospital, 1200 Main Street West, Hamilton, ON L8N 3Z5, Canada
- McMaster Immunology Research Centre, McMaster University, MDCL 4010, 1280 Main Street West, Hamilton, ON L8S 4K1, Canada
- Centre for Discovery in Cancer Research, McMaster University, MDCL 5106, 1280 Main Street West, Hamilton, ON L8S 4M1, Canada
| | - Ali A Ashkar
- Department of Medicine, McMaster University, McMaster Children's Hospital, 1200 Main Street West, Hamilton, ON L8N 3Z5, Canada
- McMaster Immunology Research Centre, McMaster University, MDCL 4010, 1280 Main Street West, Hamilton, ON L8S 4K1, Canada
- Centre for Discovery in Cancer Research, McMaster University, MDCL 5106, 1280 Main Street West, Hamilton, ON L8S 4M1, Canada
| |
Collapse
|
2
|
Qiu Z, Li Z, Liu X, Zhang R, Li Y, Gao C, Mao X, Bao Y, Zhang M, Guo C. From tumor microenvironment to emerging biomarkers: the reshaping of the esophageal squamous cell carcinoma tumor microenvironment by neoadjuvant chemotherapy combined with immunotherapy. Front Immunol 2024; 15:1478922. [PMID: 39703499 PMCID: PMC11655454 DOI: 10.3389/fimmu.2024.1478922] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2024] [Accepted: 11/15/2024] [Indexed: 12/21/2024] Open
Abstract
Esophageal squamous cell carcinoma is a cancer with high morbidity and mortality. The advent of immune checkpoint inhibitors has significantly increased complete response rates and postoperative R0 resection rates after neoadjuvant therapy. These drugs can largely reverse the suppression of the immune system caused by the tumor microenvironment, allowing the reactivation of anti-tumor immune infiltrating cells, significantly improving the patient's tumor microenvironment, and thus preventing tumor development. However, there are still some patients who respond poorly to neoadjuvant combined immunotherapy and cannot achieve the expected results. It is now found that exploring changes in the tumor microenvironment not only elucidates patient responsiveness to immunotherapy and identifies more reliable biomarkers, but also addresses the limitations of prediction with imaging examination such as CT and the instability of existing biomarkers. In light of these considerations, this review aims to delve into the alterations within the tumor microenvironment and identify potential predictive biomarkers ensuing from neoadjuvant immunotherapy in the context of esophageal squamous cell carcinoma.
Collapse
Affiliation(s)
- Zhengzhou Qiu
- Jiangxi Medical College, Nanchang University, NanChang, China
- Department of Thoracic Surgery, Jiangxi Cancer Hospital, The Second Affiliated Hospital of Nanchang Medical College, Jiangxi Cancer Institute, Nanchang, China
- Jiangxi Key Laboratory of Oncology, Jiangxi Cancer Hospital, Nanchang, Jiangxi, China
| | - Zhao Li
- Department of Thoracic Surgery, Jiangxi Cancer Hospital, The Second Affiliated Hospital of Nanchang Medical College, Jiangxi Cancer Institute, Nanchang, China
| | - Xingfei Liu
- Jiangxi Medical College, Nanchang University, NanChang, China
- Department of Thoracic Surgery, Jiangxi Cancer Hospital, The Second Affiliated Hospital of Nanchang Medical College, Jiangxi Cancer Institute, Nanchang, China
| | - Ruilin Zhang
- Jiangxi Medical College, Nanchang University, NanChang, China
- Department of Thoracic Surgery, Jiangxi Cancer Hospital, The Second Affiliated Hospital of Nanchang Medical College, Jiangxi Cancer Institute, Nanchang, China
| | - Yongxuan Li
- Jiangxi Medical College, Nanchang University, NanChang, China
- Department of Thoracic Surgery, Jiangxi Cancer Hospital, The Second Affiliated Hospital of Nanchang Medical College, Jiangxi Cancer Institute, Nanchang, China
| | - Chenggen Gao
- Jiangxi Medical College, Nanchang University, NanChang, China
- Department of Thoracic Surgery, Jiangxi Cancer Hospital, The Second Affiliated Hospital of Nanchang Medical College, Jiangxi Cancer Institute, Nanchang, China
- Jiangxi Key Laboratory of Oncology, Jiangxi Cancer Hospital, Nanchang, Jiangxi, China
| | - Xiaoling Mao
- Jiangxi Key Laboratory of Oncology, Jiangxi Cancer Hospital, Nanchang, Jiangxi, China
- Medical College, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi, China
| | - Yin Bao
- Jiangxi Key Laboratory of Oncology, Jiangxi Cancer Hospital, Nanchang, Jiangxi, China
- Medical College, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi, China
| | - Mingyue Zhang
- Jiangxi Key Laboratory of Oncology, Jiangxi Cancer Hospital, Nanchang, Jiangxi, China
- Medical College, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi, China
| | - Changying Guo
- Department of Thoracic Surgery, Jiangxi Cancer Hospital, The Second Affiliated Hospital of Nanchang Medical College, Jiangxi Cancer Institute, Nanchang, China
- Zhejiang-Jiangxi Joint Thoracic Oncology Research Laboratory, Jiangxi Cancer Hospital, Nanchang, Jiangxi, China
| |
Collapse
|
3
|
Catalano F, Brunelli M, Signori A, Rescigno P, Buti S, Galli L, Spada M, Masini C, Galuppini F, Vellone VG, Gaggero G, Maruzzo M, Merler S, Vignani F, Cavo A, Bimbatti D, Milella M, Dei Tos AP, Sbaraglia M, Murianni V, Damassi A, Cremante M, Maffezzoli M, Llaja Obispo MA, Banna GL, Fornarini G, Rebuzzi SE. Analyses of tumor microenvironment in patients with advanced renal cell carcinoma receiving immunotherapy (Meet-URO 18 study). Future Oncol 2024; 20:1495-1503. [PMID: 38682738 PMCID: PMC11441071 DOI: 10.2217/fon-2023-1068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Accepted: 04/05/2024] [Indexed: 05/01/2024] Open
Abstract
Introduction: The Meet-URO 18 study is a multicentric study of patients with metastatic renal cell carcinoma receiving nivolumab in the second-line and beyond, categorized as responders (progression-free survival ≥ 12 months) and non-responders (progression-free survival < 3 months).Areas covered: The current study includes extensive immunohistochemical analysis of T-lineage markers (CD3, CD4, CD8, CD8/CD4 ratio), macrophages (CD68), ph-mTOR, CD15 and CD56 expression on tumor cells, and PD-L1 expression, on an increased sample size including 161 tumor samples (113 patients) compared with preliminary presented data. Responders' tumor tissue (n = 90; 55.9%) was associated with lower CD4 expression (p = 0.014), higher CD56 expression (p = 0.046) and higher CD8/CD4 ratio (p = 0.030).Expert opinion/commentary: The present work suggests the regulatory role of a subpopulation of T cells on antitumor response and identifies CD56 as a putative biomarker of immunotherapy efficacy.
Collapse
Affiliation(s)
- Fabio Catalano
- Medical Oncology Unit 1, IRCCS Ospedale Policlinico San Martino, 16132, Genoa, Italy
| | - Matteo Brunelli
- Pathology Unit, Department of Diagnostics & Public Health, University & Hospital Trust of Verona, 37124, Verona, Italy
| | - Alessio Signori
- Department of Health Sciences (DISSAL), Section of Biostatistics, University of Genoa, 16132, Genoa, Italy
| | - Pasquale Rescigno
- Candiolo Cancer Institute, FPO-IRCCS, 10060, Candiolo, Italy
- Translational & Clinical Research Institute, Centre for Cancer, Newcastle University, Newcastle upon Tyne, NE1 7RU, UK
| | - Sebastiano Buti
- Department of Medicine and Surgery, University of Parma, 43126, Parma, Italy
- Medical Oncology Unit, University Hospital of Parma, 43126, Parma, Italy
| | - Luca Galli
- Medical Oncology Unit 2, Azienda Ospedaliera Universitaria Pisana, 56126, Pisa, Italy
| | - Massimiliano Spada
- UOC Oncologia Medica, Istituto Fondazione G. Giglio, 90015, Cefalù, Italy
| | - Cristina Masini
- Medical Oncology Unit, Clinical Cancer Centre, AUSL-IRCCS di Reggio Emilia, 42122, Reggio Emilia, Italy
| | - Francesca Galuppini
- Surgical Pathology Unit, Department of Medicine (DIMED), University of Padua, 35128, Padua, Italy
| | - Valerio Gaetano Vellone
- Pathology Unit, IRCCS Istituto Giannina Gaslini, 16147, Genoa, Italy
- Department of Integrated Surgical & Diagnostic Sciences (DISC), University of Genoa, 16132, Genoa, Italy
| | - Gabriele Gaggero
- Pathology Unit, IRCCS Istituto Giannina Gaslini, 16147, Genoa, Italy
| | - Marco Maruzzo
- Oncology Unit 1, Istituto Oncologico Veneto IOV–IRCCS, 35128, Padua, Italy
| | - Sara Merler
- Section of Innovation Biomedicine–Oncology Area, Department of Engineering for Innovation Medicine, University of Verona & Verona University & Hospital Trust, Verona, 37134, Italy
| | - Francesca Vignani
- Division of Medical Oncology, Ordine Mauriziano Hospital, 10128, Turin, Italy
| | - Alessia Cavo
- Oncology Unit, Villa Scassi Hospital, 16149, Genoa, Italy
| | - Davide Bimbatti
- Oncology Unit 1, Istituto Oncologico Veneto IOV–IRCCS, 35128, Padua, Italy
| | - Michele Milella
- Section of Innovation Biomedicine–Oncology Area, Department of Engineering for Innovation Medicine, University of Verona & Verona University & Hospital Trust, Verona, 37134, Italy
| | - Angelo Paolo Dei Tos
- Surgical Pathology Unit, Department of Medicine (DIMED), University of Padua, 35128, Padua, Italy
| | - Marta Sbaraglia
- Surgical Pathology Unit, Department of Medicine (DIMED), University of Padua, 35128, Padua, Italy
| | - Veronica Murianni
- Medical Oncology Unit 1, IRCCS Ospedale Policlinico San Martino, 16132, Genoa, Italy
| | - Alessandra Damassi
- Medical Oncology Unit 1, IRCCS Ospedale Policlinico San Martino, 16132, Genoa, Italy
| | - Malvina Cremante
- Medical Oncology Unit 1, IRCCS Ospedale Policlinico San Martino, 16132, Genoa, Italy
| | - Michele Maffezzoli
- Department of Medicine and Surgery, University of Parma, 43126, Parma, Italy
| | | | - Giuseppe Luigi Banna
- Department of Oncology, Portsmouth Hospitals University NHS Trust, Portsmouth, PO6 3LY, UK
- School of Pharmacy & Biomedical Sciences, University of Portsmouth, Portsmouth, PO1 2UP, UK
| | - Giuseppe Fornarini
- Medical Oncology Unit 1, IRCCS Ospedale Policlinico San Martino, 16132, Genoa, Italy
| | - Sara Elena Rebuzzi
- Medical Oncology Unit, Ospedale San Paolo, 17100, Savona, Italy
- Department of Internal Medicine & Medical Specialties (Di.M.I.), University of Genoa, 16132, Genoa, Italy
| |
Collapse
|
4
|
Rebuzzi SE, Brunelli M, Galuppini F, Vellone VG, Signori A, Catalano F, Damassi A, Gaggero G, Rescigno P, Maruzzo M, Merler S, Vignani F, Cavo A, Basso U, Milella M, Panepinto O, Mencoboni M, Sbaraglia M, Dei Tos AP, Murianni V, Cremante M, Llaja Obispo MA, Maffezzoli M, Banna GL, Buti S, Fornarini G. Characterization of Tumor and Immune Tumor Microenvironment of Primary Tumors and Metastatic Sites in Advanced Renal Cell Carcinoma Patients Based on Response to Nivolumab Immunotherapy: Preliminary Results from the Meet-URO 18 Study. Cancers (Basel) 2023; 15:cancers15082394. [PMID: 37190322 DOI: 10.3390/cancers15082394] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 04/13/2023] [Accepted: 04/19/2023] [Indexed: 05/17/2023] Open
Abstract
BACKGROUND Prognostic and predictive factors for patients with metastatic renal cell carcinoma (mRCC) treated with immunotherapy are highly warranted, and the immune tumor microenvironment (I-TME) is under investigation. METHODS The Meet-URO 18 was a multicentric retrospective study assessing the I-TME in mRCC patients treated with ≥2nd-line nivolumab, dichotomized into responders and non-responders according to progression-free survival (≥12 months and ≤3 months, respectively). The primary objective was to identify differential immunohistochemical (IHC) patterns between the two groups. Lymphocyte infiltration and the expressions of different proteins on tumor cells (CD56, CD15, CD68, and ph-mTOR) were analyzed. The expression of PD-L1 was also assessed. RESULTS A total of 116 tumor tissue samples from 84 patients (59% were primary tumors and 41% were metastases) were evaluated. Samples from responders (N = 55) were significantly associated with lower expression of CD4+ T lymphocytes and higher levels of ph-mTOR and CD56+ compared with samples from non-responders (N = 61). Responders also showed a higher CD3+ expression (p = 0.059) and CD8+/CD4+ ratio (p = 0.084). Non-responders were significantly associated with a higher percentage of clear cell histology and grading. CONCLUSIONS Differential IHC patterns between the tumors in patients who were responders and non-responders to nivolumab were identified. Further investigation with genomic analyses is planned.
Collapse
Affiliation(s)
- Sara Elena Rebuzzi
- Medical Oncology Unit, Ospedale San Paolo, 17100 Savona, Italy
- Department of Internal Medicine and Medical Specialties (Di.M.I.), University of Genoa, 16132 Genoa, Italy
| | - Matteo Brunelli
- Pathology Unit, Department of Diagnostics and Public Health, University and Hospital Trust of Verona, 37124 Verona, Italy
| | - Francesca Galuppini
- Surgical Pathology Unit, Department of Medicine (DIMED), University of Padua, 35128 Padua, Italy
| | | | - Alessio Signori
- Department of Health Sciences (DISSAL), Section of Biostatistics, University of Genoa, 16132 Genoa, Italy
| | - Fabio Catalano
- Medical Oncology Unit 1, IRCCS Ospedale Policlinico San Martino, 16132 Genoa, Italy
| | - Alessandra Damassi
- Medical Oncology Unit 1, IRCCS Ospedale Policlinico San Martino, 16132 Genoa, Italy
| | - Gabriele Gaggero
- Pathology Unit, IRCCS Istituto Giannina Gaslini, 16147 Genoa, Italy
| | - Pasquale Rescigno
- Candiolo Cancer Institute, FPO-IRCCS, 10060 Candiolo, Italy
- Translational and Clinical Research Institute, Centre for Cancer, Newcastle University, Newcastle upon Tyne NE1 7RU, UK
| | - Marco Maruzzo
- Oncology Unit 1, Istituto Oncologico Veneto IOV-IRCCS, 35128 Padua, Italy
| | - Sara Merler
- Section of Oncology, Department of Medicine, University of Verona and Verona University Hospital Trust, 37134 Verona, Italy
| | - Francesca Vignani
- Division of Medical Oncology, Ordine Mauriziano Hospital, 10128 Turin, Italy
| | - Alessia Cavo
- Oncology Unit, Villa Scassi Hospital, 16149 Genoa, Italy
| | - Umberto Basso
- Oncology Unit 1, Istituto Oncologico Veneto IOV-IRCCS, 35128 Padua, Italy
| | - Michele Milella
- Section of Oncology, Department of Medicine, University of Verona and Verona University Hospital Trust, 37134 Verona, Italy
| | - Olimpia Panepinto
- Division of Medical Oncology, Ordine Mauriziano Hospital, 10128 Turin, Italy
| | | | - Marta Sbaraglia
- Surgical Pathology Unit, Department of Medicine (DIMED), University of Padua, 35128 Padua, Italy
| | - Angelo Paolo Dei Tos
- Surgical Pathology Unit, Department of Medicine (DIMED), University of Padua, 35128 Padua, Italy
| | - Veronica Murianni
- Medical Oncology Unit 1, IRCCS Ospedale Policlinico San Martino, 16132 Genoa, Italy
| | - Malvina Cremante
- Medical Oncology Unit 1, IRCCS Ospedale Policlinico San Martino, 16132 Genoa, Italy
| | | | - Michele Maffezzoli
- Department of Medicine and Surgery, University of Parma, 43126 Parma, Italy
| | - Giuseppe Luigi Banna
- Department of Oncology, Portsmouth Hospitals University NHS Trust, Portsmouth PO6 3LY, UK
| | - Sebastiano Buti
- Department of Medicine and Surgery, University of Parma, 43126 Parma, Italy
- Medical Oncology Unit, University Hospital of Parma, 43126 Parma, Italy
| | - Giuseppe Fornarini
- Medical Oncology Unit 1, IRCCS Ospedale Policlinico San Martino, 16132 Genoa, Italy
| |
Collapse
|
5
|
Savchenko AA, Kudryavtsev IV, Isakov DV, Sadowski IS, Belenyuk VD, Borisov AG. Recombinant Human Interleukin-2 Corrects NK Cell Phenotype and Functional Activity in Patients with Post-COVID Syndrome. Pharmaceuticals (Basel) 2023; 16:ph16040537. [PMID: 37111294 PMCID: PMC10144656 DOI: 10.3390/ph16040537] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Revised: 03/31/2023] [Accepted: 04/01/2023] [Indexed: 04/07/2023] Open
Abstract
Post-COVID syndrome develops in 10–20% of people who have recovered from COVID-19 and it is characterized by impaired function of the nervous, cardiovascular, and immune systems. Previously, it was found that patients who recovered from infection with the SARS-CoV-2 virus had a decrease in the number and functional activity of NK cells. The aim of the study was to assess the effectiveness of recombinant human IL-2 (rhIL-2) administered to correct NK cell phenotype and functional activity in patients with post-COVID syndrome. Patients were examined after 3 months for acute COVID-19 of varying severity. The phenotype of the peripheral blood NK cells was studied by flow cytometry. It was found that disturbances in the cell subset composition in patients with post-COVID syndrome were characterized by low levels of mature (p = 0.001) and cytotoxic NK cells (p = 0.013), with increased release of immature NK cells (p = 0.023). Functional deficiency of NK cells in post-COVID syndrome was characterized by lowered cytotoxic activity due to the decreased count of CD57+ (p = 0.001) and CD8+ (p < 0.001) NK cells. In the treatment of patients with post-COVID syndrome with recombinant IL-2, peripheral blood NK cell count and functional potential were restored. In general, the effectiveness of using rhIL-2 in treatment of post-COVID syndrome has been proven in patients with low levels of NK cells.
Collapse
Affiliation(s)
- Andrei A. Savchenko
- Federal Research Center “Krasnoyarsk Science Center” of the Siberian Branch of the Russian Academy of Sciences, Scientific Research Institute of Medical Problems of the North, 660022 Krasnoyarsk, Russia
| | - Igor V. Kudryavtsev
- Institute of Experimental Medicine, 197376 St. Petersburg, Russia
- School of Biomedicine, Far Eastern Federal University, 690922 Vladivostok, Russia
| | - Dmitry V. Isakov
- Institute of Experimental Medicine, Pavlov First St. Petersburg State Medical University of the Russian Federation Ministry of Healthcare, 197022 St. Petersburg, Russia
| | - Ivan S. Sadowski
- Federal Research Center “Krasnoyarsk Science Center” of the Siberian Branch of the Russian Academy of Sciences, Scientific Research Institute of Medical Problems of the North, 660022 Krasnoyarsk, Russia
| | - Vasily D. Belenyuk
- Federal Research Center “Krasnoyarsk Science Center” of the Siberian Branch of the Russian Academy of Sciences, Scientific Research Institute of Medical Problems of the North, 660022 Krasnoyarsk, Russia
| | - Alexandr G. Borisov
- Federal Research Center “Krasnoyarsk Science Center” of the Siberian Branch of the Russian Academy of Sciences, Scientific Research Institute of Medical Problems of the North, 660022 Krasnoyarsk, Russia
| |
Collapse
|
6
|
Wang S, Xu G, Li M, Zheng J, Wang Y, Feng X, Luo J, Wang S, Liu H, Duan W, Zhang H, Huang D, Zhao F, Nie Y, Yang J. M1 macrophage predicted efficacy of neoadjuvant camrelizumab combined with chemotherapy vs chemotherapy alone for locally advanced ESCC: A pilot study. Front Oncol 2023; 13:1139990. [PMID: 36969032 PMCID: PMC10038194 DOI: 10.3389/fonc.2023.1139990] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2023] [Accepted: 02/20/2023] [Indexed: 03/12/2023] Open
Abstract
Introduction The efficacy and safety of immunotherapy have been widely recognized in gastrointestinal-related cancers. However, the efficacy of neoadjuvant camrelizumab for locally advanced esophageal squamous cell carcinoma (ESCC) has not been firmly established. This study compared the efficacy of camrelizumab in combination with neoadjuvant DCF (docetaxel, cisplatin and fluorouracil), with DCF alone for ESCC, and exploring biomarkers related to immune infiltration of the ESCC immunotherapy response. Methods We enrolled and randomly assigned patients with stage II-IVa ESCC to two study treatments: camrelizumab combined with docetaxel, cisplatin and fluorouracil (DCF) regimen and DCF regimen alone. The tissue for multiplex immunofluorescence (mIF) was obtained before and after neoadjuvant therapy. The Response Evaluation Criteria in Solid Tumors RECIST Version 1.1 (RECIST 1.1) and Tumor Regression Grade (TRG) was used to evaluate efficacy. Results A total of 30 patients were enrolled in the study. Following neoadjuvant camrelizumab, the objective response rate (ORR) and the disease control rate (DCR) were 46.7% (7/15) and 95.7% (14/15), respectively. No patients reported complete remission, while ORR and DCR in the chemotherapy group were 26.7% (4/15) and 86.7% (13/15), respectively. R0 resection after neoadjuvant treatment was achieved in 3 out of 15 patients in the combined group and in all patients (15/15) in the chemotherapy group. In the combined group, M1-type tumor-associated macrophages and CD56dim NK cells were more abundant in responders than in non-responders (p < 0.05). A higher M1/M2 ratio was observed in responders (p < 0.05). With respect to the NGS, among the copy number amplified genes, the 11q13 amplicon (CCND1/FGF19/FGF4/FGF3) showed the highest frequency (47%, 7/15). Conclusions Neoadjuvant camrelizumab combined with chemotherapy improved ORR in locally advanced ESCC. M1-type tumor-associated macrophages and CD56dim NK cells might be utilized to predict camrelizumab efficacy.
Collapse
Affiliation(s)
- Shu Wang
- State Key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases and Xi-jing Hospital of Digestive Diseases, Fourth Military Medical University, Xi’an, China
- Department of Digestive Surgery, Xi Jing Hospital, The Fourth Military Medical University, Xi’an, China
| | - Guanghui Xu
- State Key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases and Xi-jing Hospital of Digestive Diseases, Fourth Military Medical University, Xi’an, China
- Department of Digestive Surgery, Xi Jing Hospital, The Fourth Military Medical University, Xi’an, China
| | - Mengbin Li
- State Key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases and Xi-jing Hospital of Digestive Diseases, Fourth Military Medical University, Xi’an, China
- Department of Digestive Surgery, Xi Jing Hospital, The Fourth Military Medical University, Xi’an, China
| | - Jiyang Zheng
- State Key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases and Xi-jing Hospital of Digestive Diseases, Fourth Military Medical University, Xi’an, China
- Department of Digestive Surgery, Xi Jing Hospital, The Fourth Military Medical University, Xi’an, China
| | - Yuhao Wang
- State Key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases and Xi-jing Hospital of Digestive Diseases, Fourth Military Medical University, Xi’an, China
- Department of Digestive Surgery, Xi Jing Hospital, The Fourth Military Medical University, Xi’an, China
| | - Xiangying Feng
- State Key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases and Xi-jing Hospital of Digestive Diseases, Fourth Military Medical University, Xi’an, China
- Department of Digestive Surgery, Xi Jing Hospital, The Fourth Military Medical University, Xi’an, China
| | - Jialin Luo
- State Key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases and Xi-jing Hospital of Digestive Diseases, Fourth Military Medical University, Xi’an, China
- Department of Digestive Surgery, Xi Jing Hospital, The Fourth Military Medical University, Xi’an, China
| | - Shibo Wang
- State Key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases and Xi-jing Hospital of Digestive Diseases, Fourth Military Medical University, Xi’an, China
| | - Huan Liu
- State Key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases and Xi-jing Hospital of Digestive Diseases, Fourth Military Medical University, Xi’an, China
| | - Weiming Duan
- The Medical Department, 3D Medicines Inc., Shanghai, China
| | - Hushan Zhang
- The Medical Department, 3D Medicines Inc., Shanghai, China
| | - Depei Huang
- The Medical Department, 3D Medicines Inc., Shanghai, China
| | - Feilong Zhao
- The Medical Department, 3D Medicines Inc., Shanghai, China
| | - Yongzhan Nie
- State Key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases and Xi-jing Hospital of Digestive Diseases, Fourth Military Medical University, Xi’an, China
| | - Jianjun Yang
- State Key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases and Xi-jing Hospital of Digestive Diseases, Fourth Military Medical University, Xi’an, China
- Department of Digestive Surgery, Xi Jing Hospital, The Fourth Military Medical University, Xi’an, China
| |
Collapse
|
7
|
Han SJ, Sung N, Wang J, O'Malley BW, Lonard DM. Steroid receptor coactivator-3 inhibition generates breast cancer antitumor immune microenvironment. Breast Cancer Res 2022; 24:73. [PMID: 36316775 PMCID: PMC9620627 DOI: 10.1186/s13058-022-01568-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Accepted: 10/17/2022] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND The tumor immune microenvironment (TIME) generated by cancer-infiltrating immune cells has a crucial role in promoting or suppressing breast cancer progression. However, whether the steroid receptor coactivator-3 (SRC-3) modulates TIME to progress breast cancer is unclear. Therefore, the present study evaluates whether SRC-3 generates a tumor-promoting TIME in breast tumors using a syngeneic immune-intact mouse model of breast cancer. METHODS We employed E0771 and 4T1 breast cancer in immune-intact syngeneic female C57BL/6 and BALB/c mice, respectively. SI-2, a specific small-molecule inhibitor of SRC-3, was administered daily (2.5 mg/kg) to E0771 and 4T1 breast tumor-bearing immune-intact mice. In addition, SRC-3 knockdown (KD)-E0771 and SRC-3 KD-4T1 cells and their parental breast cancer cells were injected into their syngeneic immune-intact female mice versus immune-deficiency mice to validate that the host immune system is required for breast tumor suppression by SRC-3 KD in immune-intact mice. Furthermore, tumor-infiltrating immune cells (such as CD4+, CD8+, CD56+, and Foxp3+ cells) in E0771 and 4T1 breast cancers treated with SI-2 and in SRC-3 KD E0771 and 4T1 breast cancers were determined by immunohistochemistry. Additionally, cytokine levels in SI-2-treated and SRC-3 KD E0771 breast tumors and their control cancers were defined with a Mouse Cytokine Array. RESULTS SRC-3 inhibition by SI-2 significantly suppressed the progression of breast cancer cells (E0771 and 4T1) into breast cancers in immune-intact syngeneic female mice. SRC-3 KD-E0771 and -4T1 breast cancer cells did not produce well-developed tumors in immune-intact syngeneic female mice compared to their parental cells, but SRC-3 KD breast cancers were well developed in immune-defective host mice. SRC-3 inhibition by SI-2 and SRC-3 KD effectively increased the numbers of cytotoxic immune cells, such as CD4+ and CD8+ T cells and CD56+ NK cells, and Interferon γ (Ifng) in breast cancers compared to vehicle. However, SI-2 treatment reduced the number of tumor-infiltrating CD4+/Foxp3+ regulatory T (Treg) cells compared to vehicle treatment. In addition, SRC-3 inhibition by SI-2 and SRC-3 KD increased C-X-C motif chemokine ligand 9 (Cxcl9) expression in breast cancer to recruit C-X-C motif chemokine receptor 3 (Cxcr3)-expressing cytotoxic immune cells into breast tumors. CONCLUSIONS SRC-3 is a critical immunomodulator in breast cancer, generating a protumor immune microenvironment. SRC-3 inhibition by SI-2 or SRC-3 KD activates the Cxcl9/Cxcr3 axis in breast tumors and enhances the antitumor immune microenvironment to suppress breast cancer progression.
Collapse
Affiliation(s)
- Sang Jun Han
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, 77030, USA.
- Duncan Cancer Center for Reproductive Medicine, Baylor College of Medicine, Houston, TX, 77030, USA.
| | - Nuri Sung
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Jin Wang
- Department of Pharmacology and Chemical Biology, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Bert W O'Malley
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, 77030, USA
- Duncan Cancer Center for Reproductive Medicine, Baylor College of Medicine, Houston, TX, 77030, USA
| | - David M Lonard
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, 77030, USA.
- Duncan Cancer Center for Reproductive Medicine, Baylor College of Medicine, Houston, TX, 77030, USA.
| |
Collapse
|
8
|
Gadgeel M, AlQanber B, Buck S, Taub JW, Ravindranath Y, Savaşan S. Aberrant myelomonocytic CD56 expression in Down syndrome is frequent and not associated with leukemogenesis. Ann Hematol 2021; 100:1695-1700. [PMID: 33890142 DOI: 10.1007/s00277-021-04531-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Accepted: 04/12/2021] [Indexed: 11/29/2022]
Abstract
Children with Down syndrome (DS) are at an increased risk of developing transient abnormal myelopoiesis (TAM) and acute leukemia. Aberrant expression of CD56 has been observed on myeloid leukemic blasts in DS patients. In general, CD56 expression in acute myeloid leukemia (AML) is considered a promoter of leukemogenesis. We did a retrospective flow cytometric study to investigate mature myelomonocytic cell CD56 expression patterns in TAM, non-TAM, and leukemia cases with DS. Flow cytometric analysis showed that granulocyte and monocyte aberrant/dysplastic CD56 expression is an inherent characteristic of most DS patients irrespective of the presence of TAM or leukemia. Increased CD56 expression in monocyte and granulocyte populations in DS could be multifactorial; greater expression of RUNX1 secondary to the gene dose effect of trisomy 21 along with the maturational state of the cells are the potential contributors. Unlike AML seen in non-DS patients, CD56 overexpression in DS AML cases does not appear to play a role in leukemogenesis.
Collapse
Affiliation(s)
- Manisha Gadgeel
- Children's Hospital of Michigan, Division of Hematology/Oncology, Hematology/Oncology Flow Cytometry Laboratory, Detroit, MI, USA
| | - Batool AlQanber
- Children's Hospital of Michigan, Division of Hematology/Oncology, Hematology/Oncology Flow Cytometry Laboratory, Detroit, MI, USA
| | - Steven Buck
- Children's Hospital of Michigan, Division of Hematology/Oncology, Hematology/Oncology Flow Cytometry Laboratory, Detroit, MI, USA
| | - Jeffrey W Taub
- Children's Hospital of Michigan, Division of Hematology/Oncology, Barbara Ann Karmanos Cancer Center, Wayne State University School of Medicine, Detroit, MI, USA
| | - Yaddanapudi Ravindranath
- Children's Hospital of Michigan, Division of Hematology/Oncology, Hematology/Oncology Flow Cytometry Laboratory, Detroit, MI, USA.,Children's Hospital of Michigan, Division of Hematology/Oncology, Barbara Ann Karmanos Cancer Center, Wayne State University School of Medicine, Detroit, MI, USA
| | - Süreyya Savaşan
- Children's Hospital of Michigan, Division of Hematology/Oncology, Hematology/Oncology Flow Cytometry Laboratory, Detroit, MI, USA. .,Children's Hospital of Michigan, Division of Hematology/Oncology, Pediatric Blood and Marrow Transplant Program, Barbara Ann Karmanos Cancer Center, Central Michigan University College of Medicine, 3901 Beaubien Blvd., Detroit, MI, 48201, USA.
| |
Collapse
|
9
|
Gunesch JT, Dixon AL, Ebrahim TAM, Berrien-Elliott MM, Tatineni S, Kumar T, Hegewisch-Solloa E, Fehniger TA, Mace EM. CD56 regulates human NK cell cytotoxicity through Pyk2. eLife 2020; 9:e57346. [PMID: 32510326 PMCID: PMC7358009 DOI: 10.7554/elife.57346] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2020] [Accepted: 06/07/2020] [Indexed: 12/12/2022] Open
Abstract
Human natural killer (NK) cells are defined as CD56+CD3-. Despite its ubiquitous expression on human NK cells the role of CD56 (NCAM) in human NK cell cytotoxic function has not been defined. In non-immune cells, NCAM can induce signaling, mediate adhesion, and promote exocytosis through interactions with focal adhesion kinase (FAK). Here we demonstrate that deletion of CD56 on the NK92 cell line leads to impaired cytotoxic function. CD56-knockout (KO) cells fail to polarize during immunological synapse (IS) formation and have severely impaired exocytosis of lytic granules. Phosphorylation of the FAK family member Pyk2 at tyrosine 402 is decreased in NK92 CD56-KO cells, demonstrating a functional link between CD56 and signaling in human NK cells. Cytotoxicity, lytic granule exocytosis, and the phosphorylation of Pyk2 are rescued by the reintroduction of CD56. These data highlight a novel functional role for CD56 in stimulating exocytosis and promoting cytotoxicity in human NK cells.
Collapse
Affiliation(s)
| | - Amera L Dixon
- Baylor College of MedicineHoustonUnited States
- Rice UniversityHoustonUnited States
- Department of Pediatrics, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical CenterNew YorkUnited States
| | - Tasneem AM Ebrahim
- Department of Pediatrics, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical CenterNew YorkUnited States
- Barnard CollegeNew YorkUnited States
| | | | | | | | - Everardo Hegewisch-Solloa
- Department of Pediatrics, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical CenterNew YorkUnited States
| | - Todd A Fehniger
- Washington University School of MedicineSt. LouisUnited States
| | - Emily M Mace
- Department of Pediatrics, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical CenterNew YorkUnited States
| |
Collapse
|